Browse > Article
http://dx.doi.org/10.14348/molcells.2016.2349

The Roles of Peroxiredoxin and Thioredoxin in Hydrogen Peroxide Sensing and in Signal Transduction  

Netto, Luis E.S. (Departamento de Genetica e Biologia Evolutiva, Instituto de Biociencias, Universidade de Sao Paulo)
Antunes, Fernando (Departamento de Quimica e Bioquimica, Centro de Quimica e Bioquimica, Faculdade de Ciencias, Universidade de Lisboa)
Abstract
A challenge in the redox field is the elucidation of the molecular mechanisms, by which $H_2O_2$ mediates signal transduction in cells. This is relevant since redox pathways are disturbed in some pathologies. The transcription factor OxyR is the $H_2O_2$ sensor in bacteria, whereas Cys-based peroxidases are involved in the perception of this oxidant in eukaryotic cells. Three possible mechanisms may be involved in $H_2O_2$ signaling that are not mutually exclusive. In the simplest pathway, $H_2O_2$ signals through direct oxidation of the signaling protein, such as a phosphatase or a transcription factor. Although signaling proteins are frequently observed in the oxidized state in biological systems, in most cases their direct oxidation by $H_2O_2$ is too slow ($10^1M^{-1}s^{-1}$ range) to outcompete Cys-based peroxidases and glutathione. In some particular cellular compartments (such as vicinity of NADPH oxidases), it is possible that a signaling protein faces extremely high $H_2O_2$ concentrations, making the direct oxidation feasible. Alternatively, high $H_2O_2$ levels can hyperoxidize peroxiredoxins leading to local building up of $H_2O_2$ that then could oxidize a signaling protein (floodgate hypothesis). In a second model, $H_2O_2$ oxidizes Cys-based peroxidases that then through thiol-disulfide reshuffling would transmit the oxidized equivalents to the signaling protein. The third model of signaling is centered on the reducing substrate of Cys-based peroxidases that in most cases is thioredoxin. Is this model, peroxiredoxins would signal by modulating the thioredoxin redox status. More kinetic data is required to allow the identification of the complex network of thiol switches.
Keywords
$H_2O_2$; Peroxiredoxin; signal transduction; thiol; thiol-disulfide exchange; thioredoxin;
Citations & Related Records
연도 인용수 순위
  • Reference
1 Chae, H.Z., Chung, S.J., and Rhee, S.G. (1994).Thioredoxindependent peroxide reductase from yeast. J. Biol. Chem. 269, 27670-27678.
2 Chen, K., Kirber, M.T., Xiao, H., Yang, Y., and Keaney, J.F. (2008). Regulation of ROS signal transduction by NADPH oxidase 4 localization. J. Cell Biol. 181, 1129-1139.   DOI
3 Dagnell, M., Frijhoff, J., Pader, I., Augsten, M., Boivin, B., Xu, J., Mandal, P.K., Tonks, N.K., Hellberg, C., Conrad, M., et al. (2013). Selective activation of oxidized PTP1B by the thioredoxin system modulates PDGF-receptor tyrosine kinase signaling. Proc. Natl. Acad. Sci. USA 110, 13398-13403.   DOI
4 Day, A.M., Brown, J.D., Taylor, S.R., Rand, J.D., Morgan, B.A., and Veal, E.A. (2012). Inactivation of a peroxiredoxin by hydrogen peroxide is critical for thioredoxin-mediated repair of oxidized proteins and cell survival .Mol. Cell 45, 398-408.   DOI
5 Delaunay, A., Pflieger, D., Barrault, M. B., Vinh, J., and Toledano, M. B. (2002). A thiol peroxidase is an $H_2O_2$ receptor and redoxtransducer in gene activation. Cell 111, 471-481.   DOI
6 Denu, J.M., and Tanner, K.G. (1998). Specific and reversible inactivation of protein tyrosine phosphatases by hydrogen peroxide: evidence for a sulfenic acid intermediate and implications for redox regulation. Biochemistry 37, 5633-5642.   DOI
7 Du, Y., Zhang, H., Zhang, X., Lu, J., and Holmgren, A. (2013). Thioredoxin 1 is inactivated due to oxidation induced by peroxiredoxin under oxidative stress and reactivated by the glutaredoxin system. J. Biol. Chem. 288, 32241-32247.   DOI
8 Ferrer-Sueta, G., Manta, B., Botti, H., Radi, R., Trujillo, M., and Denicola, A. (2011). Factors affecting protein thiol reactivity and specificity in peroxide reduction. Chem. Res. Toxicol. 24, 434-450.   DOI
9 Fomenko, D.E., Koc, A., Agisheva, N., Jacobsen, M., Kaya, A., Malinouski, M., Rutherford, J.C., Siu, K.L., Jin, D.Y., Winge, D.R., et al. (2011). Thiol peroxidases mediate specific genome-wide regulation of gene expression in response to hydrogen peroxide. Proc. Natl. Acad. Sci. USA 108, 2729-2734.   DOI
10 Gutscher, M., Sobotta, M.C., Wabnitz, G.H., Ballikaya, S., Meyer, A.J., Samstag, Y., and Dick, T.P.(2009) Proximity-based protein thiol oxidation by $H_2O_2$-scavenging peroxidases. J. Biol. Chem. 284, 31532-31540.   DOI
11 Hall, A., Nelson, K., Poole, L.B., and Karplus, P.A. (2011). Structurebased insights into the catalytic power and conformational dexterity of peroxiredoxins. Antioxid. Redox Signal. 15, 795-815.   DOI
12 Hayashi, T., Ueno, Y., and Okamoto, T. (1993). Oxidoreductive regulation of nuclear factor kappa B. Involvement of a cellular reducing catalyst thioredoxin. J. Biol. Chem. 268, 11380-11388.
13 Harald, H.H.W., Schmidt, R.S., Vollbracht, C., Paulsen, G., Riley, D., Daiber, A., and Cuadrado, A. (2015). Antioxidants in translational medicine. Antioxid. Redox Signal. 10, 1130-1143.
14 Irwin, M.E., Rivera-Del Valle, N., and Chandra, J. (2013). Redox control of leukemia: from molecular mechanisms to therapeutic opportunities Antioxid. Redox Signal. 18, 1349-1383   DOI
15 Jang, H.H., Lee, K.O., Chi, Y.H., Jung, B.G., Park, S.K., Park, J.H., Lee, J.R., Lee, S.S., Moon, J.C., Yun, J. W., et al. (2004). Two enzymes in one: two yeast peroxiredoxins display oxidative stress-dependent switching from a peroxidase to a molecular chaperone function. Cell 117, 625-635   DOI
16 Jarvis, R.M., Hughes, S.M., and Ledgerwood, E.C. (2012). Peroxiredoxin 1 functions as a signal peroxidase to receive, transduce, and transmit peroxide signals in mammalian cells. Free Radic. Biol. Med. 53, 1522-1530.   DOI
17 Kwon, J., Lee, S.R., Yang, K.S., Ahn, Y., Kim, Y.J., Stadtman, E.R., and Rhee, S.G. (2004). Reversible oxidation and inactivation of the tumor suppressor PTEN in cells stimulated with peptide growth factors. Proc. Natl. Acad. Sci. USA 101, 16419-16424.   DOI
18 Jones, D.P. (2006). Redefining oxidative stress. Antioxid. Redox Signal. 8, 1865-1879.   DOI
19 Kaszubska, W., Falls, H.D., Schaefer, V.G., Haasch, D., Frost, L., Hessler, P., Kroeger, P.E., White, D.W., Jirousek, M.R., and Trevillyan, J.M. (2002). Protein tyrosine phosphatase 1B negatively regulates leptin signaling in a hypothalamic cell line. Mol. Cell. Endocrinol. 195, 109-118.   DOI
20 Krapfenbauer, K., Engidawork, E., Cairns, N., Fountoulakis, M., and Lubec, G. (2003). Aberrant expression of peroxiredoxin subtypes in neurodegenerative disorders. Brain Res. 967, 152-160.   DOI
21 Lee, S.R., Kwon, K.S., Kim, S.R., and Rhee, S.G. (1998). Reversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factor. J. Biol. Chem. 273, 15366-15372.   DOI
22 Lee, C., Lee, S.M., Mukhopadhyay, P., Kim, S.J., Lee, S.C., Ahn, W.S., Yu, M.H., Storz, G., and Ryu, S.E. (2004). Redox regulation of OxyR requires specific disulfide bond formation involving a rapid kinetic reaction path. Nat. Struct. Mol. Biol. 11, 1179-1185.   DOI
23 Lee, S., Kim, S.M., and Lee, R.T. (2013). Thioredoxin and thioredoxin target proteins: from molecular mechanisms to functional significance. Antioxid. Redox Signal. 18, 1165-1207   DOI
24 Little, C., and O'Brien, P.J. (1969). Mechanism of peroxideinactivation of the sulphydryl enzyme glyceraldehyde-3-phophate dehydrogenase. Eur. J. Biochem.10, 533-538.
25 Matthews, J.R., Wakasugi, N., Virelizier, J.L., Yodoi, J., and Hay, R.T. (1992). Thioredoxin regulates the DNA binding activity of NF-${\kappa}B$ by reduction of a disulphide bond involving cysteine 62. Nucleic Acids Res. 20, 3821-3830.   DOI
26 MacDiarmid, C.W., Taggart, J., Kerdsomboon, K., Kubisiak, M., Panascharoen, S., Schelble, K., and Eide, D.J. (2013). Peroxiredoxin chaperone activity is critical for protein homeostasis in zinc-deficient yeast. J. Biol. Chem. 288, 31313-31327.   DOI
27 Mahadev, K., Zilbering, A., Zhu, L., and Goldstein, B. J. (2001). Insulin-stimulated hydrogen peroxide reversibly inhibits proteintyrosine phosphatase 1b in vivo and enhances the early insulin action cascade. J. Biol. Chem. 276, 21938-21942.   DOI
28 Marinho, H.S., Real, C., Cyrne, L., Soares, H., and Antunes, F. (2014). Hydrogen peroxide sensing, signaling and regulation of transcription factors. Redox Biol. 2, 535-562.   DOI
29 Meng, T., Fukada, T., and Tonks, N.K. (2002). Reversible oxidation and inactivation of protein tyrosine phosphatases in vivo. Mol. Cell 9, 387-399.   DOI
30 Meyer, Y., Buchanan, B.B., Vignols, F., and Reichheld, J.P. (2009). Thioredoxins and glutaredoxins: unifying elements in redox biology. Annu. Rev. Genet. 43, 335-367.   DOI
31 Miller, E.W., Dickinson, B.C., and Chang, C.J. (2010). Aquaporin-3 mediates hydrogen peroxide uptake to regulate downstream intracellular signaling. Proc. Natl. Acad. Sci. USA 107, 15681-15686.   DOI
32 Mishina, N.M., Tyurin-Kuzmin, P.A., Markvicheva, K.N., Vorotnikov, A.V., Tkachuk, V.A., Laketa, V., et al. (2011). Does cellular hydrogen peroxide diffuse or act locally? Antioxid. Redox Signal. 14, 1-7.   DOI
33 Nystrom, T., Yang, J., and Molin, M. (2012). Peroxiredoxins, gerontogenes linking aging to genome instability and cancer. Genes Dev. 26, 2001-2008.   DOI
34 Nelson, K.J., Knutson, S.T., Soito, L., Klomsiri, C., Poole, L.B., and Fetrow, J.S. (2011). Analysis of the peroxiredoxin family: using active-site structure and sequence information for global classification and residue analysis. Proteins 79, 947-964.   DOI
35 Netto, L.E.S., Chae, H.Z., Kang, S.W., Rhee, S.G., and Stadtman, E.R. (1996). Removal of hydrogen peroxide by thiol-specific antioxidant enzyme (TSA) is involved with its antioxidant properties. TSA possesses thiol peroxidase activity. J. Biol. Chem. 271, 15315-15321.   DOI
36 Netto, L.E.S., Oliveira, M.A., Tairum-Jr, C., and da Silva Neto, J.F. (2015). Conferring specificity in redox pathways by enzymatic thiol/disulfide exchange reactions. Free Radic. Res. 16, 1-99.
37 Ogusucu, R., Rettori, D., Munhoz, D.C., Netto, L.E.S., and Augusto, O. (2007). Reactions of yeast thioredoxin peroxidases I and II with hydrogen peroxide and peroxynitrite: rate constants by competitive kinetics. Free Radic. Biol. Med. 42, 326-334.   DOI
38 Oliveira, M.A., Discola, K.F., Alves, S. V., Medrano, F. J., Guimaraes, B.G., and Netto, L.E.S. (2010). Insights into the specificity of thioredoxin reductase-thioredoxin interactions. A structural and functional investigation of the yeast thioredoxin system. Biochemistry 49, 3317-3326.   DOI
39 Palde, P.B., and Carroll, K.S. (2015). A universal entropy-driven mechanism for thioredoxin-target recognition. Proc. Natl. Acad. Sci. USA 112, 7960-7965.   DOI
40 Parsonage, D., Youngblood, D.S., Sarma, G.N., Wood, Z.A., Karplus, P.A., and Poole, L.B. (2005). Analysis of the link between enzymatic activity and oligomeric state in AhpC, a bacterial peroxiredoxin. Biochemistry 44, 10583-10592.   DOI
41 Peralta, D., Bronowska, A.K., Morgan, B., Doka, E., Van Laer, K., Nagy, P., Grater, F., and Dick, T.P. (2015). A proton relay enhances $H_2O_2$ sensitivity of GAPDH to facilitate metabolic adaptation. Nat. Chem. Biol. 11, 156-163.   DOI
42 Parsons, Z.D., and Gates, K.S. (2013). Thiol-dependent recovery of catalytic activity from oxidized protein tyrosine phosphatases. Biochemistry 52, 6412-6423.   DOI
43 Paulsen, C.E., Truong, T.H., Garcia, F.J., Homann, A., Gupta, V., Leonard, S.E., and Carrol, K.S. (2012). Peroxide-dependent sulfenylation of the EGFR catalytic site enhances kinase activity. Nat. Chem. Biol. 8, 57-64.   DOI
44 Pedroso, N., Matias, A.C., Cyrne, L., Antunes, F., Borges, C., Malho, R., de Almeida, R.F.M., Herrero, E., Marinho, H.S. (2009). Modulation of plasma membrane lipid profile and microdomains by $H_2O_2$ in Saccharomyces cerevisiae. Free Radic. Biol. Med. 46, 289-298.   DOI
45 Peskin, A.V., Low, F.M., Paton, L.N., Maghzal, G.J., Hampton, M.B., and Winterbourn, C.C. (2007). The high reactivity of peroxiredoxin 2 with $H_2O_2$ is not reflected in its reaction with other oxidants and thiol reagents. J. Biol. Chem. 282, 11885-11892.   DOI
46 Peskin, A. V., Pace, P.E., Behring, J.B., Paton, L. N., Soethoudt, M., Bachschmid, M.M., and Winterbourn, C.C. (2016). Glutathionylation of the active site cysteines of peroxiredoxin 2 and recycling by glutaredoxin. J. Biol. Chem. [Epub ahead of print]
47 Rawat, S.J., Creasy, C.L., Peterson, J.R., and Chernoff, J. (2013). The tumor suppressor Mst1 promotes changes in the cellular redox state by phosphorylation and inactivation of peroxiredoxin-1 protein. J. Biol. Chem. 288, 8762-8771.   DOI
48 Rhee, S.G., Woo, H.A., Kil, I.S., and Bae, S.H. (2012). Peroxiredoxin functions as a peroxidase and a regulator and sensor of local peroxides J. Biol. Chem. 287, 4403-4410.   DOI
49 Ragu, S., Dardalhon, M., Sharma, S., Iraqui, I, Buhagiar-Labarchede, G., Grondin, V., Kienda, G., Vernis, L, Chanet, R., Kolodner, R.D., et al. (2014). Loss of the thioredoxin reductase Trr1 suppresses the genomic instability of peroxiredoxin tsa1 mutants. PLoS One 9, e108123.   DOI
50 Rhee, S. G., and Woo, H. A. (2011) Multiple functions of peroxiredoxins:peroxidases, sensors and regulators of the intracellular messenger $H_2O_2$, and protein chaperones. Antioxid. Redox Signal. 15, 781-794.   DOI
51 Saitoh, M., Nishitoh, H., Fujii, M., Takeda, K., Tobiume, K., Sawada, Y., Kawabata, M., Miyazono, K., and Ichijo, H. (1998). Mammalian thioredoxin is a direct inhibitor of apoptosis signal‐regulating kinase (ASK). EMBO J. 17, 2596-2606.   DOI
52 Schroder, K., Zhang, M., Benkhoff, S., Mieth, A., Pliquett, R., Kosowski, J., Kruse, C., Luedike, P., Michaelis, U.R., Weissmann, N., et al. (2012). Nox4 is a protective reactive oxygen species generating vascular NADPH oxidase. Circ. Res. 110, 1217-1225.   DOI
53 Sies, H. (2014). Role of metabolic $H_2O_2$ generation: redox signaling and oxidative stress. J. Biol. Chem. 289, 8735-8741.   DOI
54 Seidler, N.W. (2013). GAPDH: Biological Properties and Diversity. Vol. 985 (Springer).
55 Sobotta, M.C., Liou, W., Stocker, S., Talwar, D., Oehler, M., Ruppert, T., Scharf, A.N., and Dick, T.P. (2015). Peroxiredoxin-2 and STAT3 form a redox relay for $H_2O_2$ signaling. Nat. Chem. Biol. 11, 64-70.   DOI
56 Toledo, J.C., Audi, R., Ogusucu, R., Monteiro, G., Netto, L.E.S., and Augusto, O. (2011). Horseradish peroxidase compound I as a tool to investigate reactive protein-cysteine residues: from quantification to kinetics. Free Radic. Biol. Med. 50, 1032-1038.   DOI
57 Tachibana, T., Okazaki, S., Murayama, A., Naganuma, A., Nomoto, A., and Kuge, S. (2009). A major peroxiredoxin-induced activation of Yap1 transcription factor is mediated by reduction sensitive disulfide bonds and reveals a low level of transcriptional activation. J. Biol. Chem. 284, 4464-4472.   DOI
58 Tairum Jr., C.A., de Oliveira, M.A., Horta, B.B., Zara, F.J., and Netto, L.E.S. (2012). Disulfide biochemistry in 2-cys peroxiredoxin: requirement of Glu50 and Arg146 for the reduction of yeast Tsa1 by thioredoxin. J. Mol. Biol. 424, 28-41.   DOI
59 Tanner, J.J., Parsons, Z.D., Cummings, A.H., Zhou, H., and Gates, K.S. (2011). Redox regulation of protein tyrosine phosphatases:structural and chemical aspects. Antioxid. Redox Signal. 15, 77-97.   DOI
60 Trujillo, M., Clippe, A., Manta, B., Ferrer-Sueta, G., Smeets, A., Declercq, J.P., Knoops, B., and Radi, R. (2007). Pre-steady state kinetic characterization of human peroxiredoxin 5: taking advantage of Trp84 fluorescence increase upon oxidation. Arch. Biochem. Biophys. 467, 95-106.   DOI
61 Turner-Ivey, B., Manevich, Y., Schulte, J., Kistner-Griffin, E., Jezierska-Drutel, A., Liu, Y., and Neumann, C.A. (2013). Role for Prdx1 as a specific sensor in redox-regulated senescence in breast cancer. Oncogene 32, 5302-5314.   DOI
62 Veal, E.A., Ross, S.J., Malakasi, P., Peacock, E., and Morgan, B.A. (2003). Ybp1 is required for the hydrogen peroxide-induced oxidation of the Yap1 transcription factor. J. Biol. Chem. 278, 30896-30904.   DOI
63 Winterbourn, C.C., and Metodiewa, D. (1999). Reactivity of biologically important thiol compounds with superoxide and hydrogen peroxide. Free Radic. Biol. Med. 27, 322-328.   DOI
64 Watson, W.H., Pohl, J., Montfort, W.R., Stuchlik, O., Reed, M.S., Powis, G., and Jones, D.P. (2003). Redox potential of human thioredoxin 1 and identification of a second dithiol/disulfide motif. J. Biol. Chem. 278, 33408-33415.   DOI
65 Winterbourn, C.C. (2008). Reconciling the chemistry and biology of reactive oxygen species. Nat. Chem. Biol. 4, 278-286.   DOI
66 Winterbourn, C.C., and Hampton, M.B. (2008). Thiol chemistry and specificity in redox signaling. Free Radic. Biol. Med. 45, 549-561.   DOI
67 Woo, H.A., Yim, S.H., Shin, D.H., Kang, D., Yu, D.Y., and Rhee, S.G. (2010). Inactivation of peroxiredoxin I by phosphorylation allows localized $H_2O_2$ accumulation for cell signaling. Cell 140, 517-528.   DOI
68 Wood, Z.A., Poole, L.B., and Karplus, P.A. (2003). Peroxiredoxin evolution and the regulation of hydrogen peroxide signaling .Science 300, 650-653.   DOI
69 Ying, J., Clavreul, N., Sethuraman, M., Adachi, T., and Cohen, R.A. (2007). Thiol oxidation in signaling and response to stress: detection and quantification of physiological and pathophysiological thiol modifications. Free Radic. Biol. Med 43, 1099-1108.   DOI
70 Abbasi, A., Corpeleijn, E., Gansevoort, R.T., Gans, R.O.B., Struck, J., Schulte, J., Hillege, H.L., van der Harst, P., Stolk, R.P., Navis, G., et al. (2014). Circulating peroxiredoxin 4 and type 2 diabetes risk: the Prevention of Renal and Vascular Endstage Disease (PREVEND) study. Diabetologia 57, 1842-1849.   DOI
71 Ahsan, M.K., Lekli, I., Ray, D., Yodoi, J., and Das, D.K. (2009). Redox regulation of cell survival by the thioredoxin superfamily: an implication of redox gene therapy in the heart Antioxid. Redox Signal. 11, 2741-2758.   DOI
72 Aslund, F., Zheng, M., Beckwith, J., and Storz, G. (1999). Regulation of the OxyR transcription factor by hydrogen peroxide and the cellular thiol-disulfide status. Proc. Natl. Acad. Sci. USA 96, 6161-6165.   DOI
73 Berndt, C., Lillig, C.H., and Holmgren, A. (2007). Thiol-based mechanisms of the thioredoxin and glutaredoxin systems: implications for diseases in the cardiovascular system. Am. J. Physiol. Heart Circ. Physiol. 292, H1227-H1236.   DOI
74 Biteau, B., Labarre, J., and Toledano, M.B. (2003). ATP-dependent reduction of cysteine-sulphinic acid by S. cerevisiae sulphiredoxin. Nature 425, 980-984.   DOI
75 Brito, P.M., and Antunes, F. (2014). Estimation of kinetic parameters related to biochemical interactions between hydrogen peroxide and signal transduction proteins. Front Chem. 2, 82.
76 Boisnard, S., Lagniel, G., Garmendia-Torres, C. Molin, M., Boy-Marcotte, E., Jacquet, M., Toledano, M.B., Labarre, J., and Chedin, S. (2009). $H_2O_2$ activates the nuclear localization of Msn2 and Maf1 through thioredoxins in Saccharomyces cerevisiae. Eukaryot. Cell 8, 1429-1438.   DOI
77 Boronat, S., Domenech, A., Paulo, E., Calvo, I.A., Garcia-Santamarina, S., Garcia, P., Encinar Del Dedo, J., Barcons, A., Serrano, E., Carmona, M., et al. (2014). Thiol-based $H_2O_2$ signalling in microbial systems. Redox Biol. 2, 395-399   DOI
78 Branco, M.R., Marinho, H.S., Cyrne, L., and Antunes, F. (2004). Decrease of $H_2O_2$ plasma membrane permeability during adaptation to $H_2O_2$ in Saccharomyces cerevisiae. J. Biol. Chem. 279, 6501-6506.   DOI
79 Brown, J.D., Day, A.M., Taylor, S.R., Tomalin, L.E., Morgan, B.A., and Veal, E.A. (2013). A peroxiredoxin promotes $H_2O_2$ signaling and oxidative stress resistance by oxidizing a thioredoxin family protein. Cell Rep. 5, 1425-1435.   DOI
80 Calvo, I.A., Boronat, S., Domenech, A., Garcia-Santamarina, S., Ayte, J., and Hidalgo, E. (2013). Dissection of a redox relay:$H_2O_2$-dependent activation of the transcription factor Pap1 through the peroxidatic Tpx1-thioredoxin cycle. Cell Rep. 5, 1413-1424.   DOI
81 Cao, J., Schulte, J., Knight, A., Leslie, N.R., Zagozdzon, A., Bronson, R., Manevich, Y., Beeson, C., and Neumann, C.A. (2009).Prdx1 inhibits tumorigenesis via regulating PTEN/AKT activity. EMBO J. 28, 1505-1517.   DOI